Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Nutr ; 131(8): 1298-1307, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38098370

RESUMEN

This study aimed to assess how Bacillus subtilis and Enterococcus faecium co-fermented feed (FF) affects the antioxidant capacity, muscle fibre types and muscle lipid profiles of finishing pigs. In this study, a total of 144 Duroc × Berkshire × Jiaxing Black finishing pigs were randomly assigned into three groups with four replicates (twelve pigs per replication). The three treatments were a basal diet (0 % FF), basal diet + 5 % FF and basal diet + 10 % FF, respectively. The experiment lasted 38 d after 4 d of acclimation. The study revealed that 10 % FF significantly increased the activity of superoxide dismutase (SOD) and catalase (CAT) compared with 0 % FF group, with mRNA levels of up-regulated antioxidant-related genes (GPX1, SOD1, SOD2 and CAT) in 10 % FF group. 10 % FF also significantly up-regulated the percentage of slow-twitch fibre and the mRNA expression of MyHC I, MyHC IIa and MyHC IIx, and slow MyHC protein expression while reducing MyHC IIb mRNA expression. Lipidomics analysis showed that 5 % FF and 10 % FF altered lipid profiles in longissimus thoracis. 10 % FF particularly led to an increase in the percentage of TAG. The Pearson correlation analysis indicated that certain molecular markers such as phosphatidic acid (PA) (49:4), Hex2Cer (d50:6), cardiolipin (CL) (72:8) and phosphatidylcholine (PC) (33:0e) could be used to indicate the characteristics of muscle fibres and were closely related to meat quality. Together, our findings suggest that 10 % FF improved antioxidant capacity, enhanced slow-twitch fibre percentage and altered muscle lipid profiles in finishing pigs.


Asunto(s)
Antioxidantes , Enterococcus faecium , Porcinos , Animales , Antioxidantes/metabolismo , Bacillus subtilis/genética , Enterococcus faecium/genética , Fibras Musculares Esqueléticas/metabolismo , ARN Mensajero/metabolismo , Lípidos
2.
J Anim Physiol Anim Nutr (Berl) ; 108(4): 1107-1118, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38567963

RESUMEN

Reducing the dietary crude protein (CP) could effectively reduce pressure on protein ingredient supplies. However, few data have been reported about the extent to which CP can be reduced and whether limiting the use of soybean meal leads to electrolyte imbalance. In this experiment, using the low protein (LP) diet [2% lower than NRC (2012)], seventy-two piglets (35 days old) were randomly divided into 2 groups with 6 replicates of 6 piglets each: CON group (CP = 18.5%) and LP group (CP = 16.5%), to investigate the effect of the LP diet on electrolyte balance, acid-base balance, intestinal structure and amino acid transport in piglets. The results revealed that the LP diet decreased the average daily gain and dietary CP digestibility, and damaged the villi structure of the small intestine. Compared with the CON diet, the potassium content decreased and the chlorine content increased in the LP diet, and similar trends were shown in piglet serum. The arterial pH, pCO2, HCO3 -, and base excess of piglets in the LP group were lower than those in the CON group, while pO2 was higher than those in the CON group. Interestingly, the LP diet significantly increased the lysine content in piglet serum and significantly decreased the levels of arginine, leucine, and glutamic acid. Furthermore, the LP diet significantly affected the expression of some amino acid transport vectors (B0AT1, EAAC1, and y+LAT1). In summary, these findings suggested that the LP diet leads to acid-base imbalance, amino acid transport disorder and amino acids imbalance in piglets, and the dietary electrolyte may be a key factor in the impact of the LP diet on piglet growth performance and intestinal health.


Asunto(s)
Equilibrio Ácido-Base , Aminoácidos , Alimentación Animal , Fenómenos Fisiológicos Nutricionales de los Animales , Dieta con Restricción de Proteínas , Animales , Porcinos/fisiología , Alimentación Animal/análisis , Dieta con Restricción de Proteínas/veterinaria , Aminoácidos/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Intestinos/fisiología , Intestinos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Dieta/veterinaria , Sistemas de Transporte de Aminoácidos/metabolismo
3.
Nucleic Acids Res ; 49(10): 5537-5552, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33999206

RESUMEN

The intestinal invasion of pathogenic microorganisms can have serious health consequences. Recent evidence has shown that the N6-methyladenosine (m6A) mRNA modification is closely associated with innate immunity; however, the underlying mechanism is poorly understood. Here, we examined the function and mechanism of m6A mRNA modification and the YTH domain-containing protein YTHDF1 (YTH N6-methyladenosine RNA-binding protein 1) in the innate immune response against bacterial pathogens in the intestine. Ribo-seq and m6A-seq analyses revealed that YTHDF1 directs the translation of Traf6 mRNA, which encodes tumor necrosis factor receptor-associated factor 6, thereby regulating the immune response via the m6A modification near the transcript's stop codon. Furthermore, we identified a unique mechanism by which the P/Q/N-rich domain in YTHDF1 interacts with the DEAD domain in the host factor DDX60, thereby regulating the intestinal immune response to bacterial infection by recognizing the target Traf6 transcript. These results provide novel insights into the mechanism by which YTHDF1 recognizes its target and reveal YTHDF1 as an important driver of the intestinal immune response, opening new avenues for developing therapeutic strategies designed to modulate the intestinal immune response to bacterial infection.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de Unión al ARN/inmunología , Animales , Células CACO-2 , Escherichia coli Enterotoxigénica/inmunología , Células Epiteliales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Porcinos , Factor 6 Asociado a Receptor de TNF/inmunología
4.
RNA Biol ; 18(4): 576-586, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32914682

RESUMEN

The production of natural antimicrobial peptides has emerged as an important mechanism of innate immunity in animals. Defensins, members of a large family of antimicrobial peptides, have been suggested as effector molecules in host defence against bacteria, fungi, protozoa and enveloped viruses. However, the molecular mechanism underlying defensin upregulation in bacterial infection remains poorly understood. The modification of mRNA by N6-adenosine methylation (m6A) on internal bases influences gene expression in eukaryotes. Here, we show that ß-defensin production triggered by Enterotoxigenic Escherichia coli K88 (E. coli K88) infection is controlled by the cellular m6A methyltransferase METTL3. Adding back with METTL3 robustly stimulated the re-expression of defensin, which further supports the conclusion. Furthermore, using a MeRIP-seq approach, we identified a functional connection between m6A dependent GPR161 signalling and the expression of defensins. Mechanistically, we found that the transcription factor FOXO6 interacted with METTL3 to trigger the transcription of GPR161 and the subsequent regulation of ß-defensin expression. The study has shed light on the mechanisms by which enterotoxigenic Escherichia coli infection promotes enteric defensin expression.


Asunto(s)
Defensinas/genética , Escherichia coli Enterotoxigénica/fisiología , Mucosa Intestinal/metabolismo , Animales , Células Cultivadas , Defensinas/metabolismo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Mucosa Intestinal/microbiología , Intestinos/microbiología , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Porcinos
5.
J Immunol ; 202(2): 567-578, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30567729

RESUMEN

A better understanding of the molecular mechanism of intestinal fatty acid absorption could lead to novel approaches to treatment and prevention of fatty acid-related metabolic diseases. Although it is confirmed that absorption of long-chain fatty acids (LCFAs) decreases during the pathological processes, the genetic basis and molecular mechanisms remain largely unknown. N 6-methyladenosine (m6A) is the most prevalent internal modification on eukaryotic mRNA. Recently, m6A has been found to play important roles in inflammation and antiviral responses. In this study, we show that deficiency of Mettl3, the core methyltransferase of m6A, exerts antimalabsorption of LCFA activity in vitro through inhibiting the inflammation response mediated by LPS. To substantiate this finding further, we found the levels of triglycerides were also sustained in cells with depleted Mettl3, which were cultured in Transwell to polarize with villus formation to simulate the situation in vivo. Mechanistically, depletion of Mettl3 decreases the m6A level of Traf6 mRNA, thereby its transcripts are entrapped in the nucleus, followed by the decreased expression of Traf6, leading to the suppression of NF-κB and MAPK signaling pathway. Thus, the inflammation response was suppressed, resulting in the sustained absorption of LCFA. Moreover, we found that ectopic expression of Traf6 largely abolishes the sustained absorption LCFA in Mettl3 depletion cells. Collectively, silencing Mettl3 could sustain LCFA absorption through blocking the TRAF6-dependent inflammation response. Our work uncovers a critical function of m6A methylation and provides insight into critical roles of Mettl3 in LCFA absorption and inflammatory disease.


Asunto(s)
Ácidos Grasos/metabolismo , Inflamación , Metiltransferasas/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/inmunología , Animales , Línea Celular , Silenciador del Gen , Absorción Intestinal , Metilación , Metiltransferasas/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/genética , Transducción de Señal , Porcinos , Factor 6 Asociado a Receptor de TNF/genética
6.
Appl Microbiol Biotechnol ; 105(4): 1657-1668, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33475796

RESUMEN

Lactic acid bacteria (LAB) play a key role in promoting health and preventing diseases because of their beneficial effects, such as antimicrobial activities, modulating immune responses, maintaining the gut epithelial barrier and antioxidant capacity. However, the mechanisms with which LAB relieve oxidative stress and intestinal injury induced by diquat in vivo are poorly understood. In the present study, Pediococcus pentosaceus ZJUAF-4 (LAB, ZJUAF-4), a selected probiotics strain with strong antioxidant capacities, was appointed to evaluate the efficiency against oxidative stress in diquat-induced intestinal injury of mice. Alanine transaminase (ALT) and aspartate aminotransferase (AST) were analyzed to estimate the liver injury. The intestinal permeability was evaluated by 4 kDa fluorescein isothiocyanate (FITC)-dextran (FD4), D-lactate (DLA), and diamine oxidase (DAO) levels. Jejunum reactive oxygen species (ROS) production was examined by dihydroethidium (DHE) staining. Western blotting was used to detect the expression of nuclear factor (erythroid-derived-2)-like 2 (Nrf2) and its downstream genes in jejunum. The gut microbiota was analyzed by high-throughput sequencing method based on the 16S rRNA genes. The results showed that ZJUAF-4 pretreatment was found to protect the intestinal barrier function and maintain intestinal redox homeostasis under diquat stimulation. Moreover, oral administration of ZJUAF-4 increased the expression of Nrf2 and its downstream genes. High-throughput sequencing analysis indicated that ZJUAF-4 contributed to restoring the gut microbiota influenced by diquat. Our results suggested that ZJUAF-4 protected the intestinal barrier from oxidative stress-induced damage by modulating the Nrf2 pathway and gut microbiota, indicating that ZJUAF-4 may have potential applications in preventing and treating oxidative stress-related intestinal diseases. KEY POINTS: • ZJUAF-4 exerted protective effects against diquat-induced intestinal injury. • Activation of Nrf2 and its downstream targets towards oxidative stress. • ZJUAF-4 administration restoring gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Intestinales , Animales , Diquat , Ratones , Estrés Oxidativo , Pediococcus pentosaceus , ARN Ribosómico 16S/genética
7.
Mediators Inflamm ; 2021: 8032125, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34158805

RESUMEN

A novel bacteriocin secreted by Clostridium butyricum ZJU-F1 was isolated using ammonium sulfate fractionation, cation exchange chromatography, affinity chromatography, and reverse-phase high-performance liquid chromatography (RP-HPLC). The bacteriocin, named CBP22, contained 22 amino acids with the sequence PSAWQITKCAGSIAWALGSGIF. Analysis of its structure and physicochemical properties indicated that CBP22 had a molecular weight of 2264.63 Da and a +1 net charge. CBP22 showed activity against E. col K88, E. coli ATCC25922, and S. aureus ATCC26923. The effects and potential mechanisms of bacteriocin CBP22 on the innate immune response were investigated with a lipopolysaccharide- (LPS-) induced mouse model. The results showed that pretreatment with CBP22 prevented LPS-induced impairment in epithelial tissues and significantly reduced serum levels of IgG, IgA, IgM, TNF-α, and sIgA. Moreover, CBP22 treatment increased the expression of the zonula occludens and reduced permeability as well as apoptosis in the jejunum in LPS-treated mice. In summary, CBP22 inhibits the intestinal injury and prevents the gut barrier dysfunction induced by LPS, suggesting the potential use of CBP22 for treating intestinal damage.


Asunto(s)
Proteínas Bacterianas/química , Bacteriocinas/química , Clostridium butyricum/metabolismo , Intestinos/lesiones , Intestinos/microbiología , Lipopolisacáridos/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Sulfato de Amonio/química , Animales , Antiinfecciosos/farmacología , Apoptosis , Escherichia coli , Hemólisis , Masculino , Ratones , Ratones Endogámicos ICR , Microesferas , Permeabilidad , Prebióticos , Staphylococcus aureus , Factor de Necrosis Tumoral alfa/sangre
8.
J Cell Physiol ; 234(7): 11227-11234, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30488492

RESUMEN

As the intestinal epithelium is vulnerable to oxidative stress because of frequent enterocyte renewal and continuous exposure to exogenous agents, it is meaningful to figure out how the epithelial cells exert antioxidant function. We previously synthesized a novel biogenic nanoselenium (BNS) particles and proved that BNS could effectively improve intestinal antioxidative function through activating Nrf2-ARE pathway. The objective of the present study was to investigate the mechanism by which BNS activate Nrf2-ARE pathway on the physiological function of intestinal epithelial cells. In the present study, we demonstrated that treatment of IPEC-J2 cells with BNS particles not only elevated the levels of downstream proteins of nuclear factor (erythroid-derived-2)-like 2 (Nrf2) such as heme oxygenase-1 and NQO-1 in a time-dependent manner which started to weaken at 12 hr after treatment but also significantly activated Nrf2, mitogen-activated protein kinase (MAPK), and protein kinase B (AKT) pathway in a time-dependent manner within 24 hr. BNS particles significantly increased the content of phosphorylated-Nrf2, without evident influence on the level of Kelch-like ECH-associated protein 1 (Keap1). Moreover, BNS also induced the activation of p38, extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase, and AKT while phosphorylating Nrf2. Using specific protein kinase inhibitors, we found that the Nrf2-phosphorylating and antioxidative effects of BNS particles were abolished when p38, ERK1/2, and AKT were significantly inhibited. Overall, our data demonstrated that BNS particles activated Nrf2-ARE pathway through p38, ERK1/2, and AKT mediated-phosphorylation of Nrf2 to improve the antioxidant function of intestinal epithelial cells.


Asunto(s)
Antioxidantes/farmacología , Mucosa Intestinal/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Selenio/farmacología , Proteínas de Transporte Vesicular/metabolismo , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Nanopartículas del Metal/química , Estrés Oxidativo/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Porcinos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Br J Nutr ; 121(11): 1255-1263, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30837028

RESUMEN

The performance of immune system is vital for defending the body from pathogens, and it plays a crucial role in health homoeostasis. In a previous study, we have shown that LFP-20, a twenty-amino acid antimicrobial peptide in the N terminus of porcine lactoferrin, modulated inflammatory response in colitis. Here, we further investigated the effects of LFP-20 on immune homoeostasis to elucidate the mechanism of its anti-inflammation action. A lipopolysaccharide (LPS)-triggered systemic inflammatory response mice model was established. On the basis of observed mucosal lesions and apoptosis in small intestine, we found increased macrophage and neutrophil infiltration in ileum after LPS stimulation. Expectedly, LFP-20 pre-treatment attenuated the LPS-mediated immune disorders in ileum. Moreover, the flow cytometry results indicated pre-treatment with LFP-20 sustained the balance of CD3+CD8+ T cells, B cells and natural killer cells in LPS-triggered immune disturbance. Simultaneously, we demonstrated LFP-20 modulated the secretion of both activated Th1-related IL-12p70, interferon-γ, TNF-α and Th2-related IL-4, IL-5 and IL-6. Furthermore, we found LFP-20 facilitated a balanced Th1 and Th2 response, which triggered cellular defence mechanisms and induced B cells to produce opsonising antibodies belonging to certain IgG subclasses to defend against LPS stimulation. Collectively, our study indicated pre-treatment with LFP-20 could defend against LPS-triggered systemic inflammatory response in mice via modulating immune homoeostasis.


Asunto(s)
Antiinflamatorios/farmacología , Homeostasis/efectos de los fármacos , Ileítis/inmunología , Inmunidad Activa/efectos de los fármacos , Lactoferrina/farmacología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ileítis/inducido químicamente , Íleon/inmunología , Lipopolisacáridos , Activación de Linfocitos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Neutrófilos/inmunología
10.
J Immunol ; 198(4): 1696-1705, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28062699

RESUMEN

Impaired epithelial barrier function disrupts immune homeostasis and increases inflammation in intestines, leading to many intestinal diseases. Cathelicidin peptides suppress intestinal inflammation and improve intestinal epithelial barrier function independently of their antimicrobial activity. In this study, we investigated the effects of Cathelicidin-WA (CWA) on intestinal epithelial barrier function, as well as the underlying mechanism, by using enterohemorrhagic Escherichia coli (EHEC)-infected mice and intestinal epithelial cells. The results showed that CWA attenuated EHEC-induced clinical symptoms and intestinal colitis, as did enrofloxacin (Enro). CWA decreased IL-6 production in the serum, jejunum, and colon of EHEC-infected mice. Additionally, CWA alleviated the EHEC-induced disruption of mucin-2 and goblet cells in the intestine. Interestingly, CWA increased the mucus layer thickness, which was associated with increasing expression of trefoil factor 3, in the jejunum of EHEC-infected mice. CWA increased the expression of tight junction proteins in the jejunum of EHEC-infected mice. Using intestinal epithelial cells and a Rac1 inhibitor in vitro, we demonstrated that the CWA-mediated increases in the tight junction proteins might depend on the Rac1 pathway. Furthermore, CWA improved the microbiota and short-chain fatty acid concentrations in the cecum of EHEC-infected mice. Although Enro and CWA had similar effects on intestinal inflammation, CWA was superior to Enro with regard to improving intestinal epithelial barrier and microbiota in the intestine. In conclusion, CWA attenuated EHEC-induced inflammation, intestinal epithelial barrier damage, and microbiota disruption in the intestine of mice, suggesting that CWA may be an effective therapy for many intestinal diseases.


Asunto(s)
Catelicidinas/farmacología , Escherichia coli Enterohemorrágica , Células Epiteliales/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Intestinos/efectos de los fármacos , Animales , Catelicidinas/uso terapéutico , Colitis/tratamiento farmacológico , Colon/citología , Colon/inmunología , Colon/microbiología , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/fisiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/fisiopatología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/fisiología , Ratones , Microbiota/efectos de los fármacos , Mucina 2/metabolismo , Neuropéptidos/metabolismo , Factor Trefoil-3/genética , Proteína de Unión al GTP rac1/metabolismo
11.
Appl Microbiol Biotechnol ; 103(15): 6231-6243, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31147754

RESUMEN

Oxidative stress plays a detrimental role in gastrointestinal disorders. Although selenium-enriched probiotics have been shown to strengthen oxidation resistance and innate immunity, the potential mechanism remains unclear. Here, we focused on the biological function of our material, selenium-enriched Bacillus paralicheniformis SR14 (Se-BP), and investigated the antioxidative effects of Se-BP and its underlying molecular mechanism in porcine jejunum epithelial cells. First, we prepared Se-BP and quantified for its selenium and bacterial contents. Then, in vitro free radical scavenging activity was measured to evaluate the potential antioxidant effect of Se-BP. Third, to induce an appropriate oxidative stress model, we adopted different concentrations of H2O2 and determined the most suitable concentration by a methyl thiazolyl tetrazolium (MTT) assay. Regarding treatment with Se-BP and H2O2, we found that Se-BP increased cell viability and prevented lactate dehydrogenase release when administered prior to H2O2 exposure. Additionally, Se-BP markedly suppressed reactive oxygen species and malondialdehyde production in cells and effectively attenuated apoptosis. Compared with incubation with H2O2 alone, treatment with Se-BP significantly promoted phosphorylation of ERK and p38 MAPK signaling molecules. When administered with ERK and p38 MAPK inhibitors, Se-BP did not alleviate the decrease in cell viability. Our results suggest that Se-BP prevents H2O2-induced cell damage by activating the ERK/p38 MAPK signaling pathways.


Asunto(s)
Antioxidantes/metabolismo , Bacillus/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Peróxido de Hidrógeno/toxicidad , Oxidantes/toxicidad , Selenio/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Especies Reactivas de Oxígeno/análisis , Transducción de Señal , Porcinos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Appl Microbiol Biotechnol ; 102(7): 2941-2948, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29453632

RESUMEN

Development of alternatives to antibiotic growth promoters (AGP) used in swine production requires a better understanding of their impacts on the gut microbiota. Supplementing fermented feed (FF) in swine diets as a novel nutritional strategy to reduce the use of AGP and feed price, can positively affect the porcine gut microbiota, thereby improving pig productivities. Previous studies have noted the potential effects of FF on the shift in benefit of the swine microbiota in different regions of the gastrointestinal tract (GIT). The positive influences of FF on swine gut microbiota may be due to the beneficial effects of both pre- and probiotics. Necessarily, some methods should be adopted to properly ferment and evaluate the feed and avoid undesired problems. In this mini-review, we mainly discuss the microbiota in both fermented feed and swine gut and how FF influences swine gut microbiota.


Asunto(s)
Alimentación Animal/microbiología , Alimentos Fermentados/microbiología , Microbioma Gastrointestinal , Microbiota/fisiología , Animales , Tracto Gastrointestinal/microbiología , Probióticos/administración & dosificación , Porcinos
13.
Biochem Biophys Rep ; 38: 101665, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38419757

RESUMEN

Pyroptosis induced by lipopolysaccharide (LPS) has an obvious impact on intestinal inflammation and immune regulation. Enterotoxigenic Escherichia coli (ETEC) K88 has been proved to induce inflammatory responses in several models, but whether E. coli K88 participates in the same process of pyroptotic cell death as LPS remains to be identified. We conducted a pilot experiment to confirm that E. coli K88, instead of Escherichia coli O157 and Salmonella typhimurium, promotes the secretion of interleukin-1 beta (IL-1ß) and interleukin-18 (IL-18) in macrophages. Further experiments were carried out to dissect the molecular mechanism both in vitro and in vivo. The Enzyme-Linked Immunosorbent Assay (ELISA) results suggested that E. coli K88 treatment increased the expression of pro-inflammatory cytokines IL-18 and IL-1ß in both C57BL/6 mice and the supernatant of J774A.1 cells. Intestinal morphology observations revealed that E. coli K88 treatment mainly induced inflammation in the colon. Real-time PCR and Western blot analysis showed that the mRNA and protein expressions of pyroptosis-related factors, such as NLRP3, ASC, and Caspase1, were significantly upregulated by E. coli K88 treatment. The RNA-seq results confirmed that the effect was associated with the activation of NLRP3, ASC, Caspase1, GSDMD, IL-18, and IL-1ß, and might also be related to inflammatory bowel disease and the tumor necrosis factor pathway. The pyroptosis-activated effect of E. coli K88 was significantly blocked by NLRP3 siRNA. Our data suggested that E. coli K88 caused inflammation by triggering pyroptosis, which provides a theoretical basis for the prevention and treatment of ETEC in intestinal infection.

14.
Anim Nutr ; 17: 408-417, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38812495

RESUMEN

A proper dietary electrolyte balance (dEB) is essential to ensure optimal growth performance of piglets. In the low-protein diet, this balance may be affected by the reduction of soybean meal and the inclusion of high levels of synthetic amino acids. The objective of this experiment was to evaluate the optimal dEB of low-protein diets and its impact on the growth performance of piglets. A total of 108 piglets (initial age of 35 d) were randomly divided into 3 groups with 6 replicates of 6 pigs each as follows: low electrolyte diet (LE group; dEB = 150 milliequivalents [mEq]/kg); medium electrolyte diet (ME group; dEB = 250 mEq/kg); high electrolyte diet (HE group; dEB = 350 mEq/kg). Results indicated that the LE and HE diet significantly decreased the average daily gain, average daily feed intake, and crude protein digestibility (P < 0.05) in piglets. Meanwhile, LE diets disrupted the structural integrity of the piglets' intestines and decreased jejunal tight junction protein (occludin and claudin-1) expression (P < 0.05). Additionally, the pH and HCO3- in the arterial blood of piglets in the LE group were lower than those in the ME and HE groups (P < 0.05). Interestingly, the LE diet significantly increased lysine content in piglet serum (P < 0.05), decreased the levels of arginine, leucine, glutamic acid, and alanine (P < 0.05), and inhibited the mammalian target of rapamycin complex 1 (mTORC1) pathway by decreasing the phosphorylation abundance of key proteins. In summary, the dietary electrolyte imbalance could inhibit the activation of the mTORC1 signaling pathway, which might be a key factor in the influence of the dEB on piglet growth performance and intestinal health. Moreover, second-order polynomial (quadratic) regression analysis showed that the optimal dEB of piglets in the low-protein diet was 250 to 265 mEq/kg.

15.
Front Microbiol ; 14: 1191392, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37789849

RESUMEN

Introduction: Recently, the research on pig intestinal microbiota has become a hot topic in the field of animal husbandry. There are few articles describing the dynamic changes of porcine fecal microbiota and metabolites at different time points from birth to market. Methods: In the present study, 381 fecal samples were collected from 633 commercial pigs at 7 time points, including the 1st day, the 10th day, the 25th day, the 45th day, the 70th day, the 120th day, and the 180th day after the birth of swine, were used for microbiome analysis by Illumina MiSeq sequencing methods while 131 fecal samples from 3 time points, the 10th day, the 25th day, and 70th day after birth, were used for metabolome analysis by LC-MS methods. Results: For the microbiome analysis, the fecal microbial richness increased over time from day 1 to 180 and the ß-diversity of fecal microbiota was separated significantly at different time points. Firmicutes were the main phyla from day 10 to 180, followed by Bacteroides. The abundance of Lactobacillus increased significantly on day 120 compared with the previous 4 time points. From day 120 to day 180, the main porcine fecal microbes were Lactobacillus, Clostridium_sensu_stricto_1, Terrisporobacter and Streptococcus. Clostridium_sensu_stricto_1 and Terrisporobacter increased over time, while Lactobacillus, Escherichia-Shigella, Lachnoclostridium decreased with the time according to the heatmap, which showed the increase or decrease in microbial abundance over time. For the metabolome analysis, the PLS-DA plot could clearly distinguish porcine fecal metabolites on day 10, 25, and 70. The most different metabolic pathways of the 3 time points were Tryptophan metabolism, Sphingolipid signaling pathway, Protein digestion and absorption. Some metabolites increased significantly over time, such as Sucrose, L-Arginine, Indole, 2,3-Pyridinedicarboxylic acid and so on, while D-Maltose, L-2-Aminoadipic acid, 2,6-diaminohexanoic acid, L-Proline were opposite. The correlation between fecal metabolites and microbiota revealed that the microbes with an increasing trend were positively correlated with the metabolites affecting the tryptophan metabolic pathway from the overall trend, while the microbes with a decreasing trend were opposite. In addition, the microbes with an increasing trend were negatively correlated with the metabolites affecting the lysine pathway. Discussion: In conclusion, this study elucidated the dynamic changes of porcine fecal microbiota and metabolites at different stages from birth to market, which may provide a reference for a comprehensive understanding of the intestinal health status of pigs at different growth stages.

16.
Sci Total Environ ; 882: 163558, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37075996

RESUMEN

High-dose ZnO is widely used to prevent diarrhea and promote growth of weaning piglets, which has led to serious problems of animal toxicity, bacterial resistance and environmental pollution. In this study, a novel alternative ZnO (AZO) was prepared and its physicochemical properties were characterized. Animal experiments were further conducted to evaluate the effects of the ZnO forms, the dose of AZO and the combinations with AZO on the growth performance, diarrhea, zinc metabolism and gut barrier function of weaning piglets. The results showed that the AZO, compared with ordinary ZnO (OZO), nano ZnO (NZO) and porous ZnO (PZO), had the largest surface area and reduced the release of Zn2+ into the gastric fluid. AZO showed better antibacterial activity on Escherichia coli K88, Staphylococcus aureus and Salmonella enteritidis but lower cytotoxicity on porcine intestinal epithelial cells. Animal experiments suggested that low-dose AZO, NZO and PZO (300 mg/kg) improved growth performance and reduced diarrhea in weaning piglets as well as high-dose OZO (3000 mg/kg). Notably, low-dose AZO had the lowest diarrhea incidence. Additionally, low-dose AZO in combination with probiotics improved digestibility and digestive enzyme activities. Low-dose AZO in combination with probiotics also upregulated the expression of the intestinal zinc transporter proteins ZIP4 and DMT1, increased zinc bioavailability, reduced faecal zinc emissions, and avoided zinc overload in the liver and oxidative damage caused by high-dose ZnO. Moreover, low-dose AZO in combination with probiotics improved the gut barrier function of weaning piglets by promoting the expression of tight junction proteins, mucins and antimicrobial peptides and increasing gut microbiota diversity and beneficial Lactobacillus. This study proposed a novel strategy to replace high-dose ZnO and antibiotics with low-dose AZO and probiotics in weaning piglets, which effectively improved growth performance and prevented diarrhea while reducing animal toxicity, bacterial resistance, heavy metal residues and zinc emission pollution.


Asunto(s)
Óxido de Zinc , Zinc , Porcinos , Animales , Zinc/toxicidad , Suplementos Dietéticos , Óxido de Zinc/química , Destete , Diarrea/veterinaria , Diarrea/microbiología , Escherichia coli , Antibacterianos
17.
Anim Nutr ; 12: 87-95, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36632618

RESUMEN

This study was conducted to investigate the effects of fermented mixed feed (FMF) on growth performance, carcass traits, meat quality, muscle amino acid and fatty acid composition and mRNA expression levels of genes related to lipid metabolism in finishing pigs. In the present study, 144 finishing pigs (Duroc × Berkshire × Jiaxing Black) were randomly allocated to 3 dietary treatments with 4 replicate pens per group and 12 pigs per pen. The dietary treatments included a basal diet (CON), a basal diet + 5% FMF and a basal diet + 10% FMF. The experiment lasted 38 d after 4 d of acclimation. The results showed that 5% and 10% FMF significantly increased the average daily gain (ADG) of the females but not the males (P < 0.05), but FMF supplementation showed no impact on carcass traits. Moreover, 10% FMF supplementation increased the meat color45 min and meat color24 h values, while it decreased the shear force relative to CON (P < 0.05). In addition, 10% FMF significantly increased the contents of flavor amino acids (FAA), total essential AA (EAA), total non-EAA (NEAA) and total AA relative to CON (P < 0.05). Furthermore, the diet supplemented with 10% FMF significantly increased the concentration of n-3 polyunsaturated fatty acids (PUFA), n-6 PUFA and total PUFA, and the PUFA to saturated fatty acids ratio (P < 0.05), suggesting that FMF supplementation increased meat quality. Moreover, compared with the CON, 10% FMF supplementation increased the mRNA expression of lipogenic genes, including CEBPα, PPARγ, SREBP1 and FABP4, and upregulated the expression of unsaturated fatty acid synthesis (ACAA1 and FADS2). Together, our results suggest that 10% FMF dietary supplementation improved the female pigs' growth performance, improved the meat quality and altered the profiles of muscle fatty acids and amino acids in finishing pigs. This study provides a reference for the production of high-quality pork.

18.
Anim Nutr ; 12: 116-127, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36632621

RESUMEN

Probiotics can improve animal growth performance and intestinal health. Bacillus species, Lactobacillus species, Bifidobacterium species, yeast etc. are the common types of probiotics. However, understanding the effects of probiotics on the immune status and gut microbiota of weaning piglets and how the probiotics exert their impact are still limited. This study aimed to investigate the effects of Bacillus amyloliquefaciens 40 (BA40) on the performance, immune status and gut microbiota of piglets. A total of 12 litters of newborn piglets were randomly divided into 3 groups. Piglets in control group were orally dosed with phosphate buffered saline; BA40 group and probiotics group were orally gavaged with resuspension BA40 and a probiotics product, respectively. The results showed that BA40 treatment significantly decreased (P < 0.05) the diarrhea incidence (from d 5 to 40), diamine oxidase, D-lactate, interleukin (IL)-1ß and interferon-γ concentrations compared with control group and probiotics group. Meanwhile BA40 dramatically increased the total antioxidant capacity, IL-10 and secretory immunoglobulin-A concentrations in contrast to control group. For the microbial composition, BA40 modulated the microbiota by improving the abundance of Bacteroides, Phascolarctobacterium (producing short-chain fatty acids) and Desulfovibrio and reducing the proliferation of pathogens (Streptococcus, Tyzzerella, Vellionella and paraeggerthella). Meanwhile, a metabolic function prediction explained that carbohydrate metabolism and amino acid metabolism enriched in BA40 group in contrast to control group and probiotics group. For correlation analysis, the results demonstrated that BA40-enriched Phascolarctobacterium and Desulfovibrio provide insights into strategies for elevating the health status and performance of weaned piglets. Altogether, BA40 exerted stronger ability in decreasing diarrhea incidence and improved antioxidant activity, gut barrier function and immune status of piglets than the other treatments. Our study provided the experimental and theoretical basis for the application of BA40 in pig production.

19.
Int J Biol Macromol ; 229: 952-963, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36596372

RESUMEN

Inulin is a typical kind of fermentable polysaccharide and has emerged as a promising dietary supplement due to its multiple health-promoting effects. This study aimed to unveil the dynamic change pattern of inulin utilizability as a fermentation substrate during gut microbiota development and illuminate its potential association with gut microbiota in Chinese Jinhua native pig models via longitudinal analyses. Herein, fresh feces were collected at one week pre- and post-weaning as well as 3rd month post-weaning, respectively. Targeted metabolomics and in vitro simulated fermentation revealed increasing concentrations of fecal short-chain fatty acids (SCFAs) and elevating utilizability of inulin as a fermentation substrate. Microbiomic analyses demonstrated the conspicuous longitudinal alteration in gut microbial composition and a significant rise in microbial community diversity during gut microbiota development. Furthermore, gut microbial functional analyses showed a remarkable increase in the relative abundances of carbohydrate metabolism pathways, including pentose phosphate pathway, galactose metabolism pathway, butanoate metabolism pathway as well as fructose and mannose metabolism pathway. Notably, relative abundances of bacterial genera Bifidobacterium, Roseburia, Faecalibacterium and Enterococcus displayed significantly positive correlations with the production of microbial fermentation-derived SCFAs. Collectively, these findings offer novel insights into understanding inulin utilizability variations from the perspective of gut microbiota development.


Asunto(s)
Microbioma Gastrointestinal , Animales , Porcinos , Inulina/metabolismo , Ácidos Grasos Volátiles/metabolismo , Bacterias/metabolismo , Heces/microbiología , Fermentación
20.
Meat Sci ; 201: 109177, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37023593

RESUMEN

This study aimed to determine the effects of fermented mixed feed (FMF) supplementation (0%, 5% and 10%) on the intestinal microbial community and metabolism, and the compositions of volatile flavor compounds and inosine monophosphate (IMP) contents in the longissimus thoracis. In this study, 144 finishing pigs (Duroc × Berkshire × Jiaxing Black) were randomly allocated to 3 groups with 4 replicate pens per group and 12 pigs per pen. The experiment lasted 38 days after 4 days of acclimation. The 16S rRNA gene sequences and an untargeted metabolomics analysis showed FMF altered the profiles of microbes and metabolites in the colon. Heracles flash GC e-nose analysis showed that 10% FMF (treatment 3) had a greater influence on the compositions of volatile flavor compounds than 5% FMF (treatment 2). Compared to 0% FMF (treatment 1), the contents of total aldehydes, (E,E)-2,4-nonadienal, dodecanal, nonanal and 2-decenal were significantly increased by treatment 3, and treatment 3 increased IMP concentrations and gene expressions related to its synthesis. Correlations analysis showed significantly different microbes and metabolites had strong correlations with the contents of IMP and volatile flavor compounds. In conclusion, treatment 3 regulated intestinal microbial community and metabolism, that in turn altered the compositions of volatile compounds, which contributed to improving pork flavor and umami.


Asunto(s)
Carne de Cerdo , Carne Roja , Animales , Metabolómica , ARN Ribosómico 16S/genética , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA